14
592 | OCTOBER 2015 | VOLUME 12 www.nature.com/nrgastro Departments of Medicine, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095‑7378, USA (E.A.M., J.S.L., K.T., S.W.C.). West Los Angeles VA Medical Center, 11301 Wilshire Boulevard, Los Angeles, CA 90073, USA (K.T.). Institute for Genomics and Bioinformatics, University of California at Irvine, 4038 Bren Hall, Irvine, CA 92697‑3435, USA (P.B.). Correspondence to: E.A.M. [email protected] Towards a systems view of IBS Emeran A. Mayer, Jennifer S. Labus, Kirsten Tillisch, Steven W. Cole and Pierre Baldi Abstract | Despite an extensive body of reported information about peripheral and central mechanisms involved in the pathophysiology of IBS symptoms, no comprehensive disease model has emerged that would guide the development of novel, effective therapies. In this Review, we will first describe novel insights into some key components of brain–gut interactions, starting with the emerging findings of distinct functional and structural brain signatures of IBS. We will then point out emerging correlations between these brain networks and genomic, gastrointestinal, immune and gut‑microbiome‑related parameters. We will incorporate this new information, as well as the reported extensive literature on various peripheral mechanisms, into a systems‑ based disease model of IBS, and discuss the implications of such a model for improved understanding of the disorder, and for the development of more‑effective treatment approaches in the future. Mayer, E. A. et al. Nat. Rev. Gastroenterol. Hepatol. 12, 592–605 (2015); published online 25 August 2015; doi:10.1038/nrgastro.2015.121 Introduction IBS is the most common functional gastrointestinal dis- order, occurring in up to 15% of the population world- wide. 1 Even though the syndrome is defined by chronically recurring abdominal pain and discomfort associated with altered bowel habits in the absence of organic disease, increased trait anxiety, as well as comorbidity with psy- chiatric and other chronic pain syndromes are common. 2 Despie an extensive body of reported information about peripheral 3–5 and central 6–9 mechanisms involved in the pathophysiology of IBS symptoms, and the development of animal models with high face and construct validity (that is, the models have many features similar to and is based on a similar pathophysiological concept as the human disease), 10 no comprehensive disease model has emerged that would guide the development of novel and effective therapies. This aspect is surprising in view of the comprehensive data as well as clinical experience demon- strating the strong relationship between psychosocial factors and IBS symptoms, 11 and in view of breakthroughs in the identification of several IBS-related biological abnormalities at several levels: the gut epithelium; 12 immune system; 4,5 neuroendocrine mechanisms; 13 brain structure and function; 7 stress response; 14 affective, 8,15 cognitive 6,16–18 and pain modulation 19,20 abnormali- ties; gene polymorphisms; 3 and the gut microbiome. 21,22 Despite these insights, a comprehensive understanding of how these various factors interact, and particularly to what degree they are involved, in the generation of symp- toms in IBS in general or in IBS subsets (as opposed to representing epiphenomena) has not emerged. The long history of IBS research 23 is full of examples of reported abnormalities (including excessive mucus production, alterations in sigmoid colon motility, slow wave abnor- malities in smooth muscle, gut inflammation and others), which were reported in small sample cohorts and often not confirmed in validation sample cohorts. Typically, the majority of reported mean differences are small when compared with healthy individuals, often do not take into account sex-related differences, might only be present in subsets of patients and correlations with clinical symptoms are weak. Thus, only a very small number of findings have translated into highly effective therapies. 24 These therapies include the serotonin (5-HT) 5-HT 3 receptor antagonist, alosetron, the 5-HT 4 agonist, tegaserod, the guanylate cyclase agonist, linaclotide, and the chloride channel blocker lubiprostone (for chronic idiopathic consti- pation). 25 Even though effective, the first two agents were restricted or withdrawn due to serious adverse effects, whereas the latter two are primarily targeted at treating the symptom of constipation. 25 The brain and gut show reciprocal interactions in health and disease. For example, altered brain outputs via the autonomic nervous system (ANS) and the hypothalamic– pituitary–adrenal (HPA) axis have been shown to influ- ence intestinal motility and secretion, 26 intestinal epithelial permeability, 3,12,27 immune function 28 and gut microbial composition. 21 In addition to these well-established influ- ences of the brain on peripheral target cells, peripheral environmental gut-directed factors—in particular, dietary factors 29 and intestinal pathogens 30 —might have an equally important role on these same processes. Regardless of primary cause for the observed peripheral findings, several of them (in particular immune and microbiota- related signalling) can feed back to the brain, setting up circular regulatory loops (Figure 1). Major barriers exist for progress towards a comprehen- sive understanding of IBS pathophysiology, which incor- porates such circular regulatory loops. Despite a wealth of information supporting all mechanisms listed already, controversy regarding the primary role of the brain versus peripheral factors has persisted in the field. The majority Competing interests The authors declare no competing interests. REVIEWS © 2015 Macmillan Publishers Limited. All rights reserved

Towards a systems view of IBS - University of Pittsburghpcpr.pitt.edu/wp-content/uploads/2018/01/Mayer-2015.pdf · IBS symptom generation in humans has not firmly been established

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Towards a systems view of IBS - University of Pittsburghpcpr.pitt.edu/wp-content/uploads/2018/01/Mayer-2015.pdf · IBS symptom generation in humans has not firmly been established

592 | OCTOBER 2015 | VOLUME 12 www.nature.com/nrgastro

Departments of Medicine, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095‑7378, USA (E.A.M., J.S.L., K.T., S.W.C.). West Los Angeles VA Medical Center, 11301 Wilshire Boulevard, Los Angeles, CA 90073, USA (K.T.). Institute for Genomics and Bioinformatics, University of California at Irvine, 4038 Bren Hall, Irvine, CA 92697‑3435, USA (P.B.).

Correspondence to: E.A.M. [email protected]

Towards a systems view of IBSEmeran A. Mayer, Jennifer S. Labus, Kirsten Tillisch, Steven W. Cole and Pierre Baldi

Abstract | Despite an extensive body of reported information about peripheral and central mechanisms involved in the pathophysiology of IBS symptoms, no comprehensive disease model has emerged that would guide the development of novel, effective therapies. In this Review, we will first describe novel insights into some key components of brain–gut interactions, starting with the emerging findings of distinct functional and structural brain signatures of IBS. We will then point out emerging correlations between these brain networks and genomic, gastrointestinal, immune and gut‑microbiome‑related parameters. We will incorporate this new information, as well as the reported extensive literature on various peripheral mechanisms, into a systems‑based disease model of IBS, and discuss the implications of such a model for improved understanding of the disorder, and for the development of more‑effective treatment approaches in the future.

Mayer, E. A. et al. Nat. Rev. Gastroenterol. Hepatol. 12, 592–605 (2015); published online 25 August 2015; doi:10.1038/nrgastro.2015.121

IntroductionIBS is the most common functional gastrointestinal dis­order, occurring in up to 15% of the population world­wide.1 Even though the syndrome is defined by chronically recurring abdominal pain and discomfort associated with altered bowel habits in the absence of organic disease, increased trait anxiety, as well as comorbid ity with psy­chiatric and other chronic pain syndromes are common.2 Despie an extensive body of reported information about peripheral3–5 and central6–9 mechanisms involved in the pathophysiology of IBS symptoms, and the development of animal models with high face and construct validity (that is, the models have many features similar to and is based on a similar patho physiological concept as the human disease),10 no compre hensive disease model has emerged that would guide the development of novel and effective therapies. This aspect is surprising in view of the comprehensive data as well as clinical experience demon­strating the strong relationship between psycho social factors and IBS symptoms,11 and in view of breakthroughs in the identi fication of several IBS­related biological abnormalities at several levels: the gut epi thelium;12 immune system;4,5 neuroendocrine mechanisms;13 brain structure and func tion;7 stress response;14 affective,8,15 cognitive6,16–18 and pain modulation19,20 abnormali­ties; gene polymorphisms;3 and the gut microbiome.21,22 Despite these insights, a comprehensive understanding of how these various factors interact, and particularly to what degree they are involved, in the generation of symp­toms in IBS in general or in IBS subsets (as opposed to representing epiphenomena) has not emerged. The long history of IBS research23 is full of examples of reported abnormalities (including excessive mucus production, alterations in sigmoid colon motility, slow wave abnor­malities in smooth muscle, gut inflammation and others),

which were reported in small sample cohorts and often not confirmed in vali dation sample cohorts. Typically, the majority of reported mean differences are small when compared with healthy individuals, often do not take into account sex­related differences, might only be present in subsets of patients and correlations with clinical symptoms are weak. Thus, only a very small number of findings have translated into highly effective therapies.24 These therapies include the serotonin (5­HT) 5­HT3 receptor antagonist, alosetron, the 5­HT4 agonist, tegaserod, the guanylate cyclase agonist, linaclotide, and the chloride channel blocker lubiprostone (for chronic idiopathic consti­pation).25 Even though effective, the first two agents were restricted or withdrawn due to serious adverse effects, whereas the latter two are primarily targeted at treating the symptom of constipation.25

The brain and gut show reciprocal interactions in health and disease. For example, altered brain outputs via the autonomic nervous system (ANS) and the hypothalamic–pituitary–adrenal (HPA) axis have been shown to influ­ence intestinal motility and secretion,26 intestinal epithelial permeability,3,12,27 immune function28 and gut microbial composition.21 In addition to these well­established influ­ences of the brain on peripheral target cells, peripheral environmental gut­directed factors—in particular, dietary factors29 and intestinal pathogens30—might have an equally important role on these same processes. Regardless of primary cause for the observed peripheral findings, several of them (in particular immune and microbiota­related signalling) can feed back to the brain, setting up circular regulatory loops (Figure 1).

Major barriers exist for progress towards a comprehen­sive understanding of IBS pathophysiology, which incor­porates such circular regulatory loops. Despite a wealth of information supporting all mechanisms listed already, controversy regarding the primary role of the brain versus peripheral factors has persisted in the field. The majority

Competing interestsThe authors declare no competing interests.

REVIEWS

© 2015 Macmillan Publishers Limited. All rights reserved

Page 2: Towards a systems view of IBS - University of Pittsburghpcpr.pitt.edu/wp-content/uploads/2018/01/Mayer-2015.pdf · IBS symptom generation in humans has not firmly been established

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 12 | OCTOBER 2015 | 593

of research and drug development have focused on single, usually peripheral, targets in preclinical models (such as ion channels or specific receptors on individual neurons). Although such studies have provided major insights into mechanisms of visceral pain,10 their relevance for IBS symptom generation in humans has not firmly been established. Integration of multiple clinical, psychosocial and biological (genetic, immune, neurobiological) find­ings into a comprehensive disease model has yet to be achieved. For example, few studies aimed at psychosocial aspects of IBS have taken neurobiological concepts into account. The focus has been on descriptive, symptom­based rather than biology­based disease definitions.

Key points

■ Physiological and molecular alterations have been identified in the brain–gut axis of human and rodent models of IBS, yet a comprehensive disease model to guide effective drug development has not emerged

■ Studies have identified distinct brain signatures in patients with IBS, which provide plausible neurobiological substrates of many previously reported behavioural and psychosocial observations

■ Emerging evidence demonstrates correlations of these brain signatures with alterations in genetics, immune system and gut microbiota in IBS, even though the causality of these interactions remains unknown

■ A systems‑biology‑based model is proposed to integrate the growing number of central, peripheral and behavioural IBS‑related alterations, and to identify targets for more effective therapies

For example, different biological mechanisms might be associated with similar clinical presentations. The com­prehensive identification of distinct biology­based sub­groups of patients (including those based on sex), with different underlying pathophysio logical components, who are differentially responsive to specific therapies has also not been achieved. A good example illustrating this point is a report on subsets of patients with IBS based on gut microbial signatures.31

In this Review, we will first describe novel insights into some key components of brain–gut–microbiome axis, starting with evolving concepts about alterations in defined structural and functional brain networks. We will discuss emerging evidence on how these brain network alterations are correlated with the immune system and the gut microbiota. We will then incorporate this infor­mation, as well as the reported extensive literature on various peripheral mechanisms, into a systems­based disease model of IBS. Rather than simply synthesizing the current knowledge about the disease, we will use this model to discuss the implications for better understanding of the disorder and for the development of more­effective treatment approaches in the future.

The nervous system componentSeveral clinical observations support a major role of the brain in IBS symptoms: the brain is ultimately respon sible for generating the subjective experience of abdom inal pain, discomfort and anxiety; stressful life events in early life have a major role in vulnerability to develop IBS, and psycho social stressors in adulthood play crucial parts in the first onset, and perceived severity of symptoms;32 centrally targeted pharmacological treatments and cog nitive behav­ioural strategies are some of the most­effective treatment strategies.33 The role of the enteric nervous system (ENS) in the regulation and coordination of motor and secretory functions of the gastrointestinal tract, in sensory function and its interactions with the brain have been extensively reviewed.34,35 Consistent with the theme of this Review, the complexity of the inter actions between multiple gut­based cell types (intrinsic and extrinsic sensory neurons, enteric glia, immune cells and inner vated entero endocrine cells) has been referred to as the ‘gut connectome’.36 The inter actions between the ENS and central nervous system (CNS), two key components of the brain–gut axis, are mediated by the sympathetic and parasympathetic branches of the ANS,6 and by multiple sensory and endo­crine pathways.34 Although it is difficult to characterize alterations of the ENS or of ANS­mediated brain–gut con­nections in living humans directly, func tional, structural and metabolic brain imaging approaches have become a highly productive avenue to study brain–gut–microbiota interactions in health and disease.8,9,37,38

Although it has long been assumed that specific brain functions such as pain processing, emotion or cogni­tion are attributable to the isolated operations of single brain regions, these processes are now viewed as result­ing from the dynamic interactions of distributed brain areas operating in large­scale networks (Figure 2). These networks and their properties have been assessed by

Nature Reviews | Gastroenterology & Hepatology

Pain, emotions, cognitions, social behaviour

Gut microbiota

Microbiomebrain loop

Brain immune loop

Microbiomeimmune loop

Brain-related mechanisms

Bowel movements

Gut-related mechanisms

■ HPA axis■ SNS

■ Cytokines■ Primed PBMCs

ENS

ANS

Microbialmetabolites

StressEarly life adversity

Social supportMedical system

DietPathogensAntibodies

Environmentalin�uences

Figure 1 | Brain–gut axis. Schematic of the brain–gut axis, including inputs from the gut microbiota, the ENS, the immune system and the external environment. The model includes both peripheral and central components, which are in bidirectional interactions. Bottom‑up influences are shown on the right side, top‑down influences on the left side of the graph. Abbreviations: ENS, enteric nervous system; HPA, hypothalamic–pituitary–adrenal; PBMC, peripheral blood mononuclear cell; SNS, sympathetic nervous system. Modified with permission from Nature Publishing Group © Irwin, M.R. & Cole, S.W. Nat. Rev. Immunol. 11, 625–632 (2011).103

REVIEWS

© 2015 Macmillan Publishers Limited. All rights reserved

Page 3: Towards a systems view of IBS - University of Pittsburghpcpr.pitt.edu/wp-content/uploads/2018/01/Mayer-2015.pdf · IBS symptom generation in humans has not firmly been established

594 | OCTOBER 2015 | VOLUME 12 www.nature.com/nrgastro

using neuro anatomical and neurophysiological studies in animals,10 as well as different brain imaging tech­niques and analy ses in humans.39–46 In humans, several types of networks have been reported: functional brain networks based on evoked responses37 or intrinsic con­nectivity of the brain during rest;39–41,44,45 structural net­works based on grey matter parameters47 and white matter properties; and anatomical networks based on white matter connecti vity.48 Both evoked and resting state studies performed in patients with IBS have dem­onstrated abnormalities in regions and task­related net­works linked to emotional arousal,49–53 central autonomic control,6,54–56 central executive control,51,57,58 sensorimotor processing6,59–61 and salience detection.57,62 IBS­related alterations in these networks have provided plausible neurobiological substrates for several information pro­cessing abnormalities reported in patients with IBS, such as biased threat appraisal and expectancy of outcomes (for example, salience network), autonomic hyperarousal (emotional arousal and central autonomic networks), and symptom­focused attention (central executive network).63

In the next section, we will discuss IBS­related changes that have been identified in these task­related networks. The individual networks are depicted in Figure 2, and their basic properties are described in Table 1.

Emotional arousal networkThis network acts as an important link between stimulus appraisal (salience network) and ANS output (generated in the central autonomic network) to peripheral targets (gastrointestinal tract, gut microbes, immune system), thereby having an important role in determining the magnitude and duration of autonomic modulation of various gut functions. A reduction in the inhibitory feed­back loop within the arousal network has been demon­strated in patients with IBS compared with healthy individuals,51,64,65 and in healthy individuals as controls after decreasing central serotonin levels by acute tryp to­phan depletion.66 Several studies published during the past decade support an increased responsiveness of emo­tional arousal circuits in relation to both expected and to delivered visceral stimuli, particularly in women.67–76 A meta­analyses of functional MRI studies published between 2000 and 2010 demonstrated that during con­trolled rectal distension, patients with IBS show more consistent activation in regions associated with emo­tional arousal than healthy individuals.53 Emotional arousal circuit reactivity is associated with 5­HT­related gene polymorphisms.77 IBS­related functional alterations are accompanied by structural brain alterations in key regions of this network.78

Nature Reviews | Gastroenterology & Hepatology

Sensorimotor network

M1

M2S2

S1

BGThal

pINSdlPFC

PPC

Central executive network

mPFC

vIPFC

pgACC

sgACC

Amyg

HippLCC

Emotional arousal network Central autonomic network

aMCC

mPFCOFC

aINSsgACC

Amyg

HypoPAG

LCC

NTSMedulla

aMCC

aINS

AmygOFC

mPFC

Salience network

Clinical symptoms resulting from the various brain network inputsBrain■ Chronic abdominal pain■ Anxiety■ Poor copingGut■ Altered bowel habits

Figure 2 | Brain networks contributing to IBS symptoms. Depicted are task‑related brain networks that have been described in the literature and for which structural and functional alterations have been reported in patients wiht IBS. The box in the centre describes the clinical symptoms related to the network inputs. Outputs most relevant for IBS pathophysiology occur in the form of descending pain modulation and autonomic nervous system activity. Abbreviations: Amyg, amygdala; aINS, anterior insula; aMCC, anterior midcingulate cortex; BG, basal ganglia; dlPFC, dorsolateral prefrontal cortex; Hipp, hippocampus; Hypo, hypothalamus; LCC, locus coeruleus complex; M1, primary motor cortex; M2, supplementary motor cortex; mPFC, medial prefrontal cortex; NTS, solitary nucleus; OFC, orbitofrontal cortex; PAG, periaqueductal grey; pgACC, pregenual anterior cingulate cortex; pINS, posteria insula; PPC, posterior parietal cortex; sgACC, subgenual anterior cingulate cortex; Thal, thalamus; vlPFC, ventrolateral prefrontal cortex.

REVIEWS

© 2015 Macmillan Publishers Limited. All rights reserved

Page 4: Towards a systems view of IBS - University of Pittsburghpcpr.pitt.edu/wp-content/uploads/2018/01/Mayer-2015.pdf · IBS symptom generation in humans has not firmly been established

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 12 | OCTOBER 2015 | 595

Central autonomic networkAn extensive body of knowledge derived from rodent studies26,79–82 supports an upregulation of brain circuits in response to chronic stress, in particular the input from the locus coeruleus complex to the amyg dala and the hypothalamus. Information from human brain imaging studies has been more limited due to the technical diffi­culties in studying key regions of this network, such as the hypothalamus, locus coeruleus complex and sub nuclei of the amygdala, primarily due to limited spatial resolution. However, pharmaco logical brain imaging studies have implicated alterations in the corticotropin­releasing factor

(also known as cortico liberin)–corticotropin­releasing receptor 164,83 and norepinephrine–α adrenergic receptor signalling system in this network.84

Sensorimotor networkAs with other chronic pain disorders,85–88 evidence indi­cates that IBS might be associated with alterations in brain networks concerned with the central processing and modulation of viscerosensory and somato sensory infor­mation. For example, compared with healthy individuals, patients with IBS showed increased low frequency power of spontaneous brain oscillations (suggesting increased

Table 1 | Brain networks with relevance to IBS

Network Core regions and inputs Key features

Default mode network

Medial prefrontal cortex, posterior cingulate or retrosplenial cortex, inferior parietal cortex, lateral temporal cortex and hippocampal formation179,180

Comprised of brain regions whose activity is greater during rest than goal‑directed task performance181

Associated with self‑related processing, including monitoring internal thoughts and future planning179,182,183

Emotional arousal network

Amygdala, its positive connections with the locus coeruleus complex and inhibitory feedback projections to the amygdala from prefrontal and anterior cingulate subregions, and from the hippocampus

Activated by perceived or real perturbation of the organism’s homeostasis

Generates rapid feedback inhibition of amygdala,184–186 thereby limiting the magnitude and duration of network activity and related activity in the central autonomic network

Central autonomic network

Control centres in the pontine‑medulla (including PAG and hypothalamus), the central nucleus of the amygdala, and several cortical regions (including the anterior INS, ACC, prefrontal and motor regions)187,188

Regions related to SNS control overlap with the executive‑processing and salience‑processing networks (including the ventral anterior INS), whereas regions related to parasympathetic control are more associated with the default mode network154

Provides central control and modulation of the ANS

Involved in regulating respiratory, cardiovascular, endocrine and digestive system activity during cognitive, affective and motor tasks and sensation

Sensorimotor network

Core cortical regions are primary somatosensory cortex (S1, post central gyrus), primary motor cortex (M1; precentral gyrus), secondary somatosensory cortex (S2) and supplemental motor area189

Close connections exist between the posterior INS (primary interoceptive cortex) and S1

Sensorimotor network connectivity to the thalamus, which relays peripheral sensory information to the cortex,190 is established by 2 years of age191

Receives sensory input from the periphery and is important for awareness of body sensations and generation of appropriate motor responses

Primary and secondary motor cortex, through their projections to the central autonomic network, might have a modulatory role in the sympathetic control of visceral function192

Central executive network

Lateral prefrontal cortices and posterior parietal cortex193

Activated during tasks involving executive functions such as attention, working memory, planning and response selection

Often coactivated with regions of the salience network,39 as the brain attempts to focus its limited processing capacity to only salient information via attention, working memory, planning and response selection193

Salience network

Dorsal ACC and anterior INS

Core regions have strong connections to medial prefrontal and temporal regions, and subcortical regions including the amygdala, PAG and basal ganglia194–196

Dorsal portions of the anterior INS receives prefrontal input, whilst ventral portions are closely linked with the amygdala and emotional arousal system

Anterior INS can be considered the main hub in the brain, switching from default mode network to activity‑related networks, and coordinating and adjusting bodily and behavioural responses to environmental changes

Responds to subjective salience of any interoceptive and exteroceptive stimulus reaching the brain, or to the expectation of such stimuli, and coordinates appropriate attentional, behavioural, affective and visceral responses to such stimuli39,197

Responds with the most appropriate responses to biologically and cognitively relevant stimuli based on maintaining homeostasis193,194 regardless of whether the subject is awake, engaged in a particular task, or asleep (achieved through close salience network connections with the other networks)

Abbreviations: ACC, anterior cingulate cortex; ANS, autonomic nervous system; INS, insula; PAG, periaqueductal grey; SNS, sympathetic nervous system.

REVIEWS

© 2015 Macmillan Publishers Limited. All rights reserved

Page 5: Towards a systems view of IBS - University of Pittsburghpcpr.pitt.edu/wp-content/uploads/2018/01/Mayer-2015.pdf · IBS symptom generation in humans has not firmly been established

596 | OCTOBER 2015 | VOLUME 12 www.nature.com/nrgastro

neural activity) in regions belonging to the sensorimotor network.59 These functional changes seem to be accom­panied by structural changes in white and grey matter. For example, patients with IBS had widespread micro­structural white matter abnormalities in sensory process­ing and/or modulation regions.89 Female patients with IBS have cortical thickness increases in sensorimotor areas that correlated with clinical measures of symptom sever­ity compared with healthy individuals.90 Such grey matter increases in patients with IBS were also seen in the poster­ior insula (INS), the primary viscerosensory cortex, and these changes were correlated with IBS symptom dura­tion.91 Volumetric grey matter analyses in a large sample of female patients with IBS revealed increases in the primary somatosensory cortex.78 Furthermore, examining struc­tural networks in IBS indicated that two key regions of the network (cingulate gyrus and thalamus) were found to be network hubs, indicating that these regions are more critical for information flow in IBS compared with health.78 When viewed together, current evidence supports the hypothesis that patients with chronically recurring vis­ceral pain and/or discomfort have functional as well as neuroplastic and microstructural alterations within the brain, particularly in regions associated with the process­ing, integration and modulation of sensory information. The mechanism(s) underlying these alterations include chronically increased viscerosensory information flow from the gut, or from dorsal horn neurons sensitized by descending pain facilitation (see Figure 3).

Central executive networkEvidence indicates that patients with IBS might have func­tional impairments in cognitive processes associated with the central executive network.16,17 Preliminary evidence based on administration of the Attentional Network Test92 suggests that patients with IBS have greater behavioural efficiency during the alerting and orienting function of attention than healthy individuals. In comparison with healthy individuals, this greater efficiency was associ­ated with greater activation of anterior midcingulate and insular cortices, confirming the previously reported close interactions between the central executive network and the salience network.39 Converging evidence also sug­gests that increased attention to gastrointestinal symp­toms and contexts have an important role in the increased percep tual sensitivity to visceral stimuli characteristic of IBS.26 Patients with IBS show deficient activation of inhibitory cortical regions involved in downregulation of pain and emotion as well as attention during expecta­tion and experi ence of aversive gastrointestinal stimuli.53 Selective recall of negative and gastrointestinal sensa­tion words, as well as selective attention to threat­related stimuli, has been demonstrated in patients with IBS.93–96 Furthermore, a reduction in the effective connectivity of the central executive network circuitry (including par­ietal, dorsal lateral prefrontal cortex) during repeated exposure to the antici pation and experience of a threat­ening gastro intestinal stimulus (repeated exposure to balloon inflations) was associated with a reduction in IBS hypersensitivity.97 Data from a sample of Japanese patients

with IBS, compared with healthy controls, indicated that alterations in error feedback mechanisms were associated with decreased dorsolateral prefrontal cortex activity.16 A strong negative correlation between the cortical thick­ness and grey matter density of the dorsolateral prefron­tal cortex and pain catastrophizing has been reported.98,99 Evidence from the University of California, Los Angeles research group indicates prepulse inhibition, a process by which an organism can filter the flow of information from its internal and external environments, is altered in IBS compared with health.60 Together, these data suggest that patients with IBS have specific abnormalities in attentional processes that have a role in the increased perception of visceral stimuli and in IBS symptom severity.

Salience networkStudies performed during the past decade on brain responses to delivered and expected rectal distension have consistently reported increased engagement of the core regions of the salience network, the anterior INS and anterior midcingulate cortex in patients with IBS,7,37 which initially but incorrectly were referred to as the “pain matrix”. In addition, a close relationship between increased affect, central emotional arousal processes and enhanced visceral stimulus perception has been reported in patients with IBS.65,100,101 Three recent reports published between 2013 and 2015 in female patients with IBS have identified disease­related alterations in anterior INS activ­ity and connectivity in the resting state58,59 and during an ambiguous abdominal pain threat,102 confirming a key role of salience network alterations in IBS. Reported altera­tions in the response and connectivity within the salience network are consistent with the prediction error character­istic of patients with IBS about the likelihood and severity of future gastrointestinal symptoms (catastrophizing).15

Integration of brain networksAn extensive body of literature supports the model of the central role of aberrant salience computation underlying key clinical IBS symptoms (depicted in Figure 3). Well­established biological and behavioural consequences exist with such a biased appraisal: the engagement of altered ANS outputs to targets in the gastrointestinal tract (inclu ding ENS activity, gut permeability, gastro intestinal motility and secretion,3 gut microbial composition and metabolites),21,22 and to extraintestinal targets (inclu ding the immune system);103 shifting of the balance of endoge­nous pain modulation systems towards increased descend­ing pain facilitation;20 increased engagement of the central executive network resulting in selective attention to gastro­intestinal symptoms; and development of prediction errors about likelihood and severity of symptoms (so­called catastrophizing).104,105 The model also provides a plausible bio logical basis for the effectiveness of different behav­ioural interventions such as cognitive behavioural therapy (normalizing salience, executive control and emotional arousal networks), self­relaxation techniques (normalizing emotional arousal and central autonomic networks) and mindfulness­based stress reduction (normalizing salience and executive control networks).63

REVIEWS

© 2015 Macmillan Publishers Limited. All rights reserved

Page 6: Towards a systems view of IBS - University of Pittsburghpcpr.pitt.edu/wp-content/uploads/2018/01/Mayer-2015.pdf · IBS symptom generation in humans has not firmly been established

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 12 | OCTOBER 2015 | 597

Genetics and epigeneticsAs with many other chronic diseases that involve the brain, IBS probably has a strong developmental compo­nent, which starts with the interactions of genetic and epi­genetic factors early in life, including the prenatal period. In addition to the reported associations of gene variations with various peripheral mechanisms,106 imaging genetics studies performed in large samples of well­phenotyped individuals with and without IBS have identified inter­actions between early environmental factors,107 candi date gene polymorphisms and brain networks related to emo­tional arousal and/or central autonomic control, salience and somatosensory integration. The reported genes were related to the regulation of the HPA axis: corticotropin­releasing hormone receptor 1 (CRHR1, single nucleo­tide polymorphisms [SNPs] rs7209436, rs110402 and rs242924); glucocorticoid receptor gene (NR3C1, SNPs rs2963155 and rs33389);108,109 female sex hormones (progesterone receptor or PGR, SNPs rs1042838 and rs10895068);109 5­HT signalling system (HTR3A c.–42C>T SNP rs1062613),77 inflammation­related genes (IL1B, SNP rs16944);108 and catecholaminergic signalling (ADRA1D, SNP rs1556832; ADRA2B, SNP rs1042717; COMT, SNP rs174697).110 As discussed in the next section, these inter­actions are markedly influenced by epigenetic factors111,112

(such as a history of early adverse life events [EALs]) and by the sex of the study participant (Figure 4).

As with other polygenic disorders, it has become clear that no single gene variation is sufficient to explain the full clinical phenotype in IBS. However, interactions between multiple genes, early life experiences and sex probably make a small contribution to the overall vari­ance of the peripheral and central endophenotypes.107 Validation studies in larger samples are required to confirm such contributions.

The external environment componentVarious influences originating in the external environ­ment (Figure 1) can markedly affect the development, chronicity and severity of IBS. Although some of these influences are mediated by the brain (psychosocial stress, social support, societal responses to symptoms) others are mediated by the gastrointestinal tract, includ­ing the gut microbiota, diet, gastrointestinal infections and medications.

Brain-mediated influencesClinical literature on the role of psychosocial factors in the development, symptom persistence and symptom flares in IBS is extensive.18 In addition to the well­documented

Nature Reviews | Gastroenterology & Hepatology

Computation of salienceProcessing of visceral signals and

generation of outputs

Generation of conscious ‘gut feeling’Integration of salience in the human insular

cortex forms a global emotional moment

Contextual cues Cognition, perceptionEmotion

Target-speci�cANS response

based onsalience

assessment

Regional gastrointestinal transit, gut permeability,microbial composition and metabolites, immune function Pain and discomfortAltered bowel habits

Descending spinalmodulation based

on salience

Posterior Anterior

dACCrACCHypo

Amyg

dIPFCIns

orbFC

PAG

A6

RVM

+ –

aINS

Progressive remapping of visceral afferent signal andintegration with cognitive, motivational, affective and

memory-related inputs

Figure 3 | Cross‑sectional integrated brain–gut model of IBS pathophysiology. Proposed model for involvement of brain–gut axis in the generation of cardinal IBS symptoms (chronic abdominal pain associated with altered bowel habits). Under normal circumstances, visceral and external signals are evaluated by the salience network, which generates brain outputs in terms of targeted ANS responses (regulating gastrointestinal and immune function) and descending pain modulatory activity (regulating pain sensitivity at the dorsal horn level). Target organ alterations (either peripherally or ANS stimulated) are signalled back to the brain via neural, endocrine or immune‑related channels. These signals are processed within subregions of the INS, and depending on their subjective salience, are consciously perceived (associated with activation of anterior INS) as normal gut sensations, discomfort or pain. IBS symptoms can arise from several primary peripheral or central mechanisms, but once brain–gut interactions are altered, causality is difficult to determine. Abbreviations: Amyg, amygdala; ANS, autonomic nervous system; dACC, dorsal anterior cingulate cortex; dlPFC, dorsolateral prefrontal cortex; Hypo, hypothalamus; INS, insula; orbFC, orbitofrontal cortex; PAG, periaqueductal grey; rACC, rostral anterior cingulate cortex; RVM, rostral ventromedial medulla.

REVIEWS

© 2015 Macmillan Publishers Limited. All rights reserved

Page 7: Towards a systems view of IBS - University of Pittsburghpcpr.pitt.edu/wp-content/uploads/2018/01/Mayer-2015.pdf · IBS symptom generation in humans has not firmly been established

598 | OCTOBER 2015 | VOLUME 12 www.nature.com/nrgastro

role of stressful life events in adults in preceding or exacerbating IBS symptoms,32 a history of EALs is associ­ated with an increased vulnerability to IBS,113 disorders of mood and affect,114,115 as well as to a wide range of other chronic diseases.116 In contrast to the earlier emphasis on the role of a sexual abuse history, it is now clear that a variety of factors that disturb the quality of the inter­actions between the primary care giver and the child during the first 18 years of life (including serious illness of the mother, marital discord, divorce, verbal and emo­tional abuse) can have equally detrimental effect on adult disease vulnerability.116

Evidence from brain imaging studies have identified structural and functional brain alterations associated with self­reports of EALs. Structural alterations in regions of the emotional arousal circuitry78 and in regions associ­ated with modulating somatosensory and viscerosensory processes90 were correlated with such reports. Alterations in the activity in the brainstem and amygdala in response to noradrenergic stimulation has been associated with increasing levels of EALs, consistent with an upregu lation of central autonomic circuits in IBS.117 Self­reports of EALs were also found to be correlated with brain acti vity in net­works involved in determining the salience of somatic, visceral or environmental stimuli in IBS.57 These results suggest that the experience of adversity early in life can lead to altered resting state activity in the salience and in the central executive network of adults with IBS, possibly leading to permanent alterations in salience computation

of viscerosensory signals by the brain. The observed alterations in the functional connectivity of the emotional arousal51 and salience networks57 found in IBS might be driven by altered central noradrenergic modulation.50,64 Similar changes have also been observed in animals exposed to early life stressors,118 and have been linked to increased sympathetic nervous system responses.119 EALs are also associated with altered signalling within the HPA axis.120,121 A study published in 2009 demonstrated that self­report of EALs was associated with exaggerated HPA axis responses to an aversive visceral stimulus, an effect that was more pronounced in male participants.113

Considerable preclinical and clinical evidence supports the concept that gene expression can be influenced by EALs through epigenetic mechanisms, including DNA methylation, and that these effects can persist throughout adult life.115,122,123 As depicted in Figure 4, interactions of EALs, sex and vulnerability gene polymorphisms might increase the risk of developing IBS by shaping the connec­tivity of relevant brain networks (see previous section). Even though the prevalence, clinical importance and underlying molecular mechanisms of EALs have been studied in great detail, there are other factors through which the external environment can influence brain func­tion. These include, but are not limited to, the beneficial role of a strong social support system, which can miti­gate the negative effects of EALs, and societal responses to patients’ symptom reporting, which can drive a vicious cycle of symptom amplification.124

Nature Reviews | Gastroenterology & Hepatology

PFC

OFCAM

sACC

HF

Early adverse life events

Brain geneexpression

Gut gene expression

Epigenetic factors,including microbial products

Sex

Race

Microbial metabolitesImmune cells

Stress and allostatic load Intermediate brainphenotypes

Clinicalphenotype

Executivenetwork

Saliencenetwork

Sensori-motor

network

Emotional arousal or central

autonomicnetwork

IBS

Epithelialpermeability

Neuro-muscularsystem

Bile acidmetabolism

Immunefunction

Intermediate gutphenotypes

Figure 4 | Longitudinal brain–gut model of IBS pathophysiology. The interaction between genetic and epigenetic influences result in central and peripheral gene expression profiles that underlie the shaping of brain‑based and gut‑based intermediate phenotypes. Epigenetic factors provide the input from environmental influences on the development of intermediate phenotypes. Brain and gut intermediate phenotypes interact bidirectionally to shape the clinical phenotype of IBS. Abbreviations: AM, amygdala; HF, hippocampal formation; OFC, orbitofrontal cortex; PFC, prefrontal cortex; sACC, subgenual anterior cingulate cortex.

REVIEWS

© 2015 Macmillan Publishers Limited. All rights reserved

Page 8: Towards a systems view of IBS - University of Pittsburghpcpr.pitt.edu/wp-content/uploads/2018/01/Mayer-2015.pdf · IBS symptom generation in humans has not firmly been established

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 12 | OCTOBER 2015 | 599

Gastrointestinal-tract-mediated influencesFactors arising from the external environment have been implicated in the pathophysiology of IBS and in the modu­lation of IBS symptoms. These factors have been reviewed elsewhere and so will not be discussed in detail here: dietary factors;125–128 pathogenic micro organisms;129,130 and antibiotic treatment.131,132 It remains to be determined if patients with IBS have abnormal mucosal responses to any of these factors, if symptoms reported in relation to these factors are mediated by alter ations in the gut micro­biota (see section on gut micro biota), or if it is simply the sensitivity of visceral perception that determines if some­body develops symptoms or can tolerate the same factors without any symptoms.

The immune system componentIBS is not an inflammatory disease, but a growing body of research suggests that dysregulation in immune function might nevertheless contribute to its aetiology or symp­toms.4,5,133 Mixed data exist on whether plasma or intestinal mucosal cytokine levels are associated with IBS.5 However, other research has linked IBS to an increased reactivity of blood monocytes and increased numbers of mucosal mast cells.134–139 Some studies also indicate that peripheral blood mononuclear cells (PBMCs) from patients with IBS show abnormal release of proinflammatory cytokines such as IL­6, IL­1b and TNF.5

These observations in IBS are consistent with the broader role of the nervous system in regulating immune cell development and gene expression via neuro­endocrine signals from the brain (for example, corti­sol from the HPA) and activation of sympathetic nerve fibres in the one marrow and other lymphoid organs (for example, via the sympathetic neuroeffector mol­ecule norepin ephrine).103,140–142 These regulatory interac­tions enable the CNS to integrate information regarding the general internal state of the body with information regarding real and perceived environmental threats (as detected by the salience network) and historical or devel­opmental influences (for example, a history of EALs143). As one example of these regulatory effects, studies have found that stressful life circumstances are associated with the activation of a so­called conserved transcrip­tional response to adversity (CTRA) in PBMCs that is characterized by increased expression of proinflamma­tory genes and decreased expression of genes involved in innate anti viral responses (for example, type I inter­ferons) and IgG antibody production.103,140,144,145 CTRA gene expression can also be experimentally invoked by social stress in animal models145,146 and is mediated in part by sym pathetic nervous system (SNS)­induced increases in bone marrow haematopoietic production of imma­ture and immuno logically primed monocytes (CD16– in humans, Ly­6chi in mice).146 These primed monocytes can also be reciprocally recruited into the CNS by exposure of mice to social threat and aggression.147 The integration of these observations suggests a new hypothesis regard­ing the immune system’s role in the pathogenesis of IBS: high levels of SNS acti vity during early developmental periods (stemming from either genetic or environmental

triggers such as EALs) might lead to increased production of immature primed monocytes that both traffic into the gut to alter local function and ENS plasticity, and traffic into the brain to affect CNS plasticity (including struc­tures involved in salience processing and autonomic regulation). The result can be viewed as a physiological Hebbian association between gut biology and brain func­tion, resulting in a self­perpetuating feedback system in which a sensitized gut generates ongoing adverse sensory experi ences (symptom flares), to which a neurally sen­sitized brain responds with both greater aversion and increased sympathetic outflow, resulting in upregulated monocyte production that further promotes neural alterations in both the gut and the brain.

Several areas of empirical evidence are consistent with this systems­level brain–immune–gut hypothesis, inclu­ding: brain regulation of primed monocyte production via the SNS;148 stress­induced migration of primed mono­cytes to the brain;147 inflammation­induced neuro plastic changes in the brain149 (for example, altering affective behaviour147); reinstatement of previous stress effect on brain and behaviour by subsequent exposure to mild stress weeks after the initial sensitization;148 and the observa­tion that patients with IBS (and animal models of EAL) show increased stress responsiveness (for example, SNS activity150) and increased responsiveness of brain cir­cuits related to salience detection, emotional arousal and autonomic response.151,152

Additional evidence was examined from a pilot study in which the PBMC gene expression profiles from 20 patients with IBS (12 female) and 20 healthy indi­viduals (nine female).153 Analyses identified 280 gene transcripts showing >10% differential expression across groups (134 genes upregulated in PBMC from IBS, and 146 downregulated). Promoter­based bioinformatics analysis implicated several transcription control path­ways in structuring the observed transcriptome differ­ences, including increased activity of CREB transcription factors (which mediate β­adrenergic signalling from the SNS), growth control pathways (for example, the MAPK­responsive transcription factor ELK1), oxidative stress response pathways (NRF2), and pathways involved in growth factor and cytokine signalling (STAT). However, PBMCs derived from patients with IBS did not differ from those from healthy individuals in the activity of proinflammatory transcription factors such as NFκB or AP­1. Transcript origin analyses154 indicated that IBS upregulated genes derived predominately from mono­cytes and dendritic cells. Additional transcriptome repre­sentation analyses suggest that these effects stemmed at least in part from upregulation of immature (CD16–) monocytes within the PBMC pool of patients with IBS. These results are all consistent with a pattern of increased myeloid lineage cell development in patients with IBS, which might stem from tonically increased SNS signalling to bone marrow myelopoietic processes.146

Consistent with the hypothesis that CNS processes might mediate relationships between IBS and periph­eral myelopoiesis, differential signalling by the myeloid lineage transcription factor MZF­1 was positively

REVIEWS

© 2015 Macmillan Publishers Limited. All rights reserved

Page 9: Towards a systems view of IBS - University of Pittsburghpcpr.pitt.edu/wp-content/uploads/2018/01/Mayer-2015.pdf · IBS symptom generation in humans has not firmly been established

600 | OCTOBER 2015 | VOLUME 12 www.nature.com/nrgastro

associated with the morphometry of brain regions of the emotional arousal network155 (previously reported to be altered in IBS78,90,156). These associations included bilat­eral amygdala, hippocampal and anterior INS volumes, and bilateral anterior INS cortical thickness (all P <0.05). Moreover, these associations were reasonably specific in that other brain regions showed either no associ­ation, or negative associations (for example, bilateral cerebellar volumes).

In conjunction with the published data reviewed here, these preliminary results suggest that key regions of the emotional arousal and/or central autonomic net­works, which differ structurally between IBS and healthy indivi duals are also related to differential gene regula­tion in the peripheral immune system. We hypothesize that these chronic influences of the brain on peripheral monocytes (‘top down’) might have a role in the reported PBMC abnormalities in IBS, whereas primed monocytes migrating to the brain might have a role in the (‘bottom up’) generation of visceral hypersensitivity, anxiety and neuroplasticity during recurrent stressors.

The gut microbiota componentEvidence from rodent studies supporting bidirectional interactions between the gut microbiota and the nervous system (both CNS and ENS) has been summarized in numerous review articles.157–160 Even though various sig­nalling mechanisms underlying such interactions have been proposed, detailed mechanistic studies regarding the relative contributions of neural, hormonal, metabolite or immune­mediated factors are required to draw definitive conclusions. Evidence from human studies using different endpoints (symptoms, brain imaging studies) confirming the rodent findings, or identifying a definitive pattern of dysbiosis in patients with IBS are limited.158

Conflicting evidence exists regarding alterations to the organization and function of the gut micro­biota in patients with chronic abdominal pain and in adult and paediatric patients with IBS.21,22 Several studies examining faecal samples from patients with IBS reported decreased proportions of the genera Bifidobacterium and Lactobacillus, and increased ratios of Firmicutes:Bacteroidetes at the phylum level, even though a causal role of these microbial changes in clini­cal symptoms has not been established. On the other hand, one might speculate that some of these changes might be related to alterations in regional gut transit and secretion secondary to altered ANS output. A study in a cohort of well­phenotyped patients with IBS high­lights the complexities of brain–gut–microbiota altera­tions in IBS.31 IBS subgroups were identified based on hierarchical clustering of operational taxonomic unit information from 16S ribosomal RNA analyses.31 Two IBS clusters were clearly separated from each other, from a ‘normal­like’ IBS sample and from the healthy control sample. Although the normal­like IBS sample showed normal diversity and normal Firmicutes:Bacteroidetes ratio (as in, similar to the pattern in the healthy control sample), another cluster showed diminished, and a third cluster showed increased diversity. Both of these clusters

showed increased Firmicutes:Bacteroidetes ratios. Only the normal­like IBS group showed a markedly elevated rate of depression symptoms (40%). In addition, pro­longed colonic transit times, common in patients with depression correlated with the prevalence of 17 taxa.31 In a randomized, placebo­controlled study of healthy men and women, psychological distress and anxiety improved after taking a Lactobacillus-containing and Bifidobacterium­containing probiotic compared to those taking a matched control product, although another study using a different Lactobacillius probiotic failed to confirm these findings.161,162

Findings from brain imaging studies in healthy indi­viduals and patients with IBS has provided some evi­dence supporting reported rodent gut microbiota–brain interactions. One study has shown that chronic ingestion of a probiotic consortium (Bifididobacterium animalis ssp. lactis, Lactococcus lactis spp. lactis, Lactobacillus delbrueckii spp. bulgaricus and Streptococcus thermo-philus) for 4 weeks altered functional brain responses to an emotional face recognition task in healthy women.52 Compared with two control groups (one group received a nonfermented milk product and the other no treatment), the women who had ingested the probiotic had a reduced response to the task across a wide network of brain regions that included sensory and emotional regions. Although no treatment­related changes in self­report of symptoms of anxiety or depression were seen, the find­ings suggest a basic change in responsiveness to negative emotional stimuli in the environment. No organi zational changes in the gut microbiota were observed in this and a previous nonimaging study using the same interven­tion.163 The effects of the probiotic intervention on brain responsiveness were probably mediated by a change in microbial­derived metabolites; this hypothesis will have to be confirmed in future studies. Preliminary evidence investigating correlations between gut micro bial metabo­lites and brain structure in healthy indivi duals and patients with IBS demonstrated statistically significant correlations between several metabolites and structural aspects of several brain regions.153

An integrated modelHypothesisOn the evidence reviewed here, the following model is proposed that lends itself to experimental evaluation. The hypersensitive brain, and presumably ENS, shows increased responses to a variety of viscero sensory and exterosensory stimuli, which by themselves might not be consciously perceived in healthy individuals or in patients with inflammatory bowel disorders in whom intact descending inhibitory bulbospinal influences reduce dorsal horn excitability.20,164 This hyper­responsiveness might be a primary genetic or epigenetic alteration in certain brain networks (autonomic, emotional arousal or salience), or might be secondary to chronic experience of increased sensory input from the gut. Such increased viscerosensory signalling could originate from any of the elements of the gut connectome. Altered brain networks generate altered signals, which are transmitted to the

REVIEWS

© 2015 Macmillan Publishers Limited. All rights reserved

Page 10: Towards a systems view of IBS - University of Pittsburghpcpr.pitt.edu/wp-content/uploads/2018/01/Mayer-2015.pdf · IBS symptom generation in humans has not firmly been established

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 12 | OCTOBER 2015 | 601

periphery through the ANS, HPA axis and descending modulation to the dorsal horn. The chronically increased ANS output, results in an extensive remodelling of various peripheral cells in the immune system, ENS, gut epithe­lium (permeability), and in the composition and function of the gut microbiota, all contributing to sensi tization of visceral afferent pathways, and increased viscero sensory feedback to the brain, reinforcing the circular regulatory loops. Memory formations at the level of the immune system, the nervous system and the gut microbiota early in life are likely to contribute to the chronicity of symptoms.

Systems biology viewThe evidence reviewed above strongly suggests that IBS is a systems disease (Figure 5), involving not only complex individual systems—nervous, immune, digestive, micro­biota and the environment—but also their complex, non­linear, reciprocal interactions. Thus, we propose a systems biological view of IBS pathophysiology, an approach taken in other areas of biology and disease165,166 involving a large number of functionally diverse, interacting components, each of which contributes only a small fraction to the vari­ance of the symptoms. These components interact highly selectively at multiple scales and typically in a nonlinear fashion to produce coherent behaviours and outcomes.167 Applied on a macroscopic level, we propose this model to characterize the interactions of the nervous system, immune system, the gut environment (motility and

secretion), the gut microbiota and the external environ­ment (Figures 1 and 5). The nonlinearity of the model is reflected by the complexity of these macroscopic com­ponents and their macroscopic interactions, which form circular regulatory loops. Applied to the microscopic level, the model proposes the interactions between highly diverse cells types making up the various macroscopic components (Figure 5).

Given the complexity of the expanded gut–microbiota–brain–environment axis, and with the rapid advance of ana­lytical tools, in particular of ‘­omics’ technologies—in the case of IBS studies, primarily geno mics, transcripto mics, proteomics, metabo lomics, brain connectomics and their integration into, for instance, gene and protein networks—we believe that systems bio logical approaches are essential to fully understand symptom generation and to identify novel treatment approaches in the future.

The multiorgan, systemic view of IBS leads to the pre­diction that disease models focusing on individual cellu­lar components at the macroscopic level (brain, immune system, gut, microbiota, environment) will have limited validity. Furthermore, this view predicts that therapies targeted at single organs (for example, brain, immune system, gut), single mechanisms (for example, secretion, motility, gut microbiota, pain, diet), or single molecular targets (such as ion channels or receptors) are not likely to be successful in treating the entire syndrome, as evidenced by the limited success in drug development for IBS to date, reflected in rather small effect sizes on overall changes in IBS symptoms. This problem is compounded by the fact that different subsets of patients can be charac terized by different patterns of interactions between the components of the overall system, resulting in differential responses to a particular therapy.

Indeed, each isolated mechanism proposed over the past 30 years only explains a small fraction of the vari­ance of the clinical phenotype26 and no isolated therapies have had better effectiveness than ~10% over placebo.168 In addition, the systems view predicts that a single agent targeting multi ple components of the system (tricyclic anti depressants, 5­HT3 antagonists), or combined thera­pies, aimed at simultaneously targeting multiple organs and mechanisms (combination of laxatives, anti diarrhoeal agents, probiotics and centrally acting drugs) are likely to be more effective in the clinic, than individual treatments by themselves. Although no controlled studies have been conducted yet and are needed, in our opinion, this sug­gested integrated approach is in part supported by current clinical practices and the consensus of most physicians.

Although still in their infancy, integrated multiomic approaches could be essential in the future to better under­stand the disease spectrum for IBS at the system level and the underlying mechanisms. Ideally, one would like to combine high­throughput microscopy and brain imaging to obtain brain connectome variables, and sequencing and mass spectrometry methods to obtain complete measurements of genomic, epi genomic, transcriptomic, proteomic and metabolomic variables simultaneously in multiple organs and tissues (in particular in gut and brain), including the micro biota. Data derived from these

O

O

O

O

O

O

O

O

O

O

*O

*O

O

O–

O–

O–

R

*NH3

H

HO

CH4

CH4

H

H

H2C

H2C

Nature Reviews | Gastroenterology & Hepatology

Brain networks

Transcriptional network

Microbiota

Immunecells

Noncoding RNA networkProtein network

Metabolite network

Figure 5 | Systems biological model of IBS. Schematic illustrating a systems biological view of components involved in the development of IBS at the cellular and molecular level. Systems‑based interactions between central and peripheral components of the brain–gut axis can be studied at the level of the genome, epigenome, transcriptome, proteome, metabolome and brain connectome.

REVIEWS

© 2015 Macmillan Publishers Limited. All rights reserved

Page 11: Towards a systems view of IBS - University of Pittsburghpcpr.pitt.edu/wp-content/uploads/2018/01/Mayer-2015.pdf · IBS symptom generation in humans has not firmly been established

602 | OCTOBER 2015 | VOLUME 12 www.nature.com/nrgastro

–omics approaches can be integratively analysed in com­bination with other sources of information ranging from public databases to the liter ature to identify, for instance, targetable hubs. This integrated –omics approach faces major obstacles stemming from the obvious difficulties of performing some of these analyses in human patients (for example, nervous system tissue). However, impor­tant steps can be taken in this direction both using rodent models10,81,169,170 and by performing partial measurements in humans.

Some of these analyses should be performed at multiple time points along the circadian cycle. Circadian rhythms are found at the molecular level in all tissues, both centrally and peripherally, and have a fundamental role in coordin­ating the physiology and homeo stasis of the organism. Indeed integrative system biology analyses171 have revealed that about 10% of all transcripts and metabo lites oscillate in a circadian manner in any tissue,172 with little overlap across different tissues beyond the core molecular clock comprising a transcriptional–translational negative feed­back loop coordinated by a dozen genes.173 Furthermore, the list of oscillating molecular species in a given tissue is altered by genetic, epigenetic and environmental pertur­bations and thus provides a characteristic physiological signature of a tissue and its health state.174–176 Furthermore, complex reciprocal interactions exist between the mol­ecular rhythms found in different brain regions and in other organs. Thus, in short, it is reasonable to predict that IBS should result in perturbed lists of transcripts and metabolites that oscillate in a circadian manner both centrally in specific brain areas as well as peripherally, for instance in the gut.

ConclusionsClearly, none of the growing list of individual abnormali­ties identified in patients with IBS by itself can account for the variance of the clinical phenotype. For the same reason, such individual abnormalities are unlikely to rep­resent reliable biomarkers and are not likely to represent suitable targets for the development of highly effective treatments. Rather, as depicted schematically in Figures 1 and 5, the clinical phenotype emerges from the inter­actions of multiple systems in the periphery (gut con­nectome, microbiome, genome and epigenome) and in the brain (connectome) interacting with each other in bidirectional ways. Most consistent with a systems view is the concept that central and peripheral abnor­malities form circular loops that reinforce each other. In the absence of comprehensive phenotyping studies in patients with IBS performed longitudinally with or without therapeutic interventions, and without targeted mechanistic animal studies, it remains unclear which of these reported abnormalities are primary and which are secondary. On the basis of this assessment, we suggest that in the future, high­throughput –omics measurement across both tissues and time, combined with comprehen­sive characterization of clinical, behavioural and brain endophenotypes, should enable more accurate differen­tial analyses, uncover complex system­level interactions and, ultimately, help develop more efficient, multi­pronged therapies.177 Such a system biological approach might not only hold promise when applied to IBS and related functional gastrointestinal disorders, but also for inflammatory diseases of the gut, such as IBD and coeliac disease.178

1. Longstreth, G. F. et al. in ROME III: The Functional Gastrointestinal Disorders (eds Drossman, D. A. et al.) 487–556 (Degnon Associates, 2006).

2. Mayer, E. A. & Bushnell, M. C. in Functional Pain Syndromes: Presentation and Pathophysiology (eds Mayer, E. A. & Bushnell, M. C.) 531–565 (IASP Press, 2009).

3. Camilleri, M. Peripheral mechanisms in irritable bowel syndrome. N. Engl. J. Med. 367, 1626–1635 (2012).

4. Hughes, P. A. et al. Immune activation in irritable bowel syndrome: can neuroimmune interactions explain symptoms? Am. J. Gastroenterol. 108, 1066–1074 (2013).

5. Ohman, L. & Simren, M. Pathogenesis of IBS: role of inflammation, immunity and neuroimmune interactions. Nat. Rev. Gastroenterol. Hepatol. 7, 163–173 (2010).

6. Fukudo, S. Stress and visceral pain: focusing on irritable bowel syndrome. Pain 154 (Suppl. 1), S63–S70 (2013).

7. Mayer, E. A., Gupta, A., Kilpatrick, L. K. & Hong, J. Y. Chronic visceral pain. Pain 156 (Suppl. 1), S50–S63 (2015).

8. Elsenbruch, S. Abdominal pain in irritable bowel syndrome: a review of putative psychological, neural and neuro‑immune mechanisms. Brain Behav. Immun. 25, 386–394 (2011).

9. Al Omran, Y. & Aziz, Q. Functional brain imaging in gastroenterology: to new beginnings. Nat. Rev. Gastroenterol. Hepatol. 11, 565–576 (2014).

10. Greenwood‑Van Meerveld, B., Prusator, D. K. & Johnson, A. C. Animal models of visceral pain:

pathophysiology, translational relevance and challenges. Am. J. Physiol. Gastrointest. Liver Physiol. 308 , G885–G903 (2015).

11. Lackner, J. M. in Functional and GI Motility Disorders (eds Quigley, E. M. M., Hongo, M. & Fukudo, S.) 104–116 (Karger Medical and Scientific Publishers, 2014).

12. Bischoff, S. C. et al. Intestinal permeability —a new target for disease prevention and therapy. BMC Gastroenterol. 14, 189 (2014).

13. Mawe, G. M. & Hoffman, J. M. Serotonin signalling in the gut—functions, dysfunctions and therapeutic targets. Nat. Rev. Gastroenterol. Hepatol. 10, 473–486 (2013).

14. Larauche, M., Mulak, A. & Tache, Y. Stress and visceral pain: from animal models to clinical therapies. Exp. Neurol. 233, 49–67 (2012).

15. Elsenbruch, S. How positive and negative expectations shape the experience of visceral pain. Handb. Exp. Pharmacol. 225, 97–119 (2014).

16. Aizawa, E. et al. Altered cognitive function of prefrontal cortex during error feedback in patients with irritable bowel syndrome, based on FMRI and dynamic causal modeling. Gastroenterology 143, 1188–1198 (2012).

17. Kennedy, P. J. et al. Cognitive performance in irritable bowel syndrome: evidence of a stress‑related impairment in visuospatial memory. Psychol. Med. 44, 1553–1566 (2014).

18. Tanaka, Y., Kanazawa, M., Fukudo, S. & Drossman, D. A. Biopsychosocial model of irritable bowel syndrome. J. Neurogastroenterol. Motil. 17, 131–139 (2011).

19. Piche, M., Arsenault, M., Poitras, P., Rainville, P. & Bouin, M. Widespread hypersensitivity is related to altered pain inhibition processes in irritable bowel syndrome. Pain 148, 49–58 (2010).

20. Wilder‑Smith, C. H. The balancing act: endogenous modulation of pain in functional gastrointestinal disorders. Gut 60, 1589–1599 (2011).

21. Mayer, E. A., Savidge, T. & Shulman, R. J. Brain–gut microbiome interactions and functional bowel disorders. Gastroenterology 146, 1500–1512 (2014).

22. Simren, M. et al. Intestinal microbiota in functional bowel disorders: a Rome foundation report. Gut 62, 159–176 (2013).

23. Drossman, D. A. in The Growth of Gastroenterologic Knowledge in the 20th Century (ed. Kirsner, J. B.) 419–432 (Lea & Febiger, 1993).

24. Mayer, E. A., Tillisch, K. & Bradesi, S. Review article: modulation of the brain‑gut axis as a therapeutic approach in gastrointestinal disease. Aliment. Pharmacol. Ther. 24, 919–933 (2006).

25. Barboza, J. L., Talley, N. J. & Moshiree, B. Current and emerging pharmacotherapeutic options for irritable bowel syndrome. Drugs 74, 1849–1870 (2014).

26. Mayer, E. A. et al. Functional GI disorders: from animal models to drug development. Gut 57, 384–404 (2008).

27. Keita, A. V. & Soderholm, J. D. The intestinal barrier and its regulation by neuroimmune factors. Neurogastroenterol. Motil. 22, 718–733 (2010).

REVIEWS

© 2015 Macmillan Publishers Limited. All rights reserved

Page 12: Towards a systems view of IBS - University of Pittsburghpcpr.pitt.edu/wp-content/uploads/2018/01/Mayer-2015.pdf · IBS symptom generation in humans has not firmly been established

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 12 | OCTOBER 2015 | 603

28. Elenkov, I. J. & Chrousos, G. P. Stress system—organization, physiology and immunoregulation. Neuroimmunomodulation 13, 257–267 (2006).

29. Hayes, P. A., Fraher, M. H. & Quigley, E. M. Irritable bowel syndrome: the role of food in pathogenesis and management. Gastroenterol. Hepatol. (N. Y.) 10, 164–174 (2014).

30. Spiller, R. & Lam, C. An Update on post‑infectious irritable bowel syndrome: role of genetics, immune activation, serotonin and altered microbiome. J. Neurogastroenterol. Motil. 18, 258–268 (2012).

31. Jeffery, I. B. et al. An irritable bowel syndrome subtype defined by species‑specific alterations in faecal microbiota. Gut 61, 997–1006 (2012).

32. Mayer, E. A. The neurobiology of stress and gastrointestinal disease. Gut 47, 861–869 (2000).

33. Ford, A. C. et al. Effect of antidepressants and psychological therapies, including hypnotherapy, in irritable bowel syndrome: systematic review and meta‑analysis. Am. J. Gastroenterol. 109, 1350–1365 (2014).

34. Mayer, E. A. Gut feelings: the emerging biology of gut–brain communication. Nat. Rev. Neurosci. 12, 453–466 (2011).

35. Furness, J. B. The enteric nervous system and neurogastroenterology. Nat. Rev. Gastroenterol. Hepatol. 9, 286–294 (2012).

36. Bohorquez, D. V. & Liddle, R. A. The gut connectome: making sense of what you eat. J. Clin. Invest. 125, 888–890 (2015).

37. Mayer, E. A. et al. Brain imaging approaches to the study of functional GI disorders: a Rome working team report. Neurogastroenterol. Motil. 21, 579–596 (2009).

38. Farmer, A. D., Aziz, Q., Tack, J. & Van Oudenhove, L. The future of neuroscientific research in functional gastrointestinal disorders: integration towards multidimensional (visceral) pain endophenotypes? J. Psychosom. Res. 68, 475–481 (2010).

39. Seeley, W. W. et al. Dissociable intrinsic connectivity networks for salience processing and executive control. J. Neurosci. 27, 2349–2356 (2007).

40. Guo, C. C. et al. One‑year test‑retest reliability of intrinsic connectivity network fMRI in older adults. Neuroimage 61, 1471–1483 (2012).

41. Bajaj, S., Adhikari, B. M. & Dhamala, M. Higher frequency network activity flow predicts lower frequency node activity in intrinsic low‑frequency BOLD fluctuations. PLoS ONE 8, e64466 (2013).

42. Bullmore, E. & Sporns, O. The economy of brain network organization. Nat. Rev. Neurosci. 13, 336–349 (2012).

43. Rubinov, M. & Sporns, O. Complex network measures of brain connectivity: uses and interpretations. Neuroimage 52, 1059–1069 (2010).

44. Hutchison, R. M. et al. Resting‑state connectivity identifies distinct functional networks in macaque cingulate cortex. Cereb. Cortex 22, 1294–1308 (2012).

45. Greicius, M. D., Supekar, K., Menon, V. & Dougherty, R. F. Resting‑state functional connectivity reflects structural connectivity in the default mode network. Cereb. Cortex 19, 72–78 (2009).

46. Sporns, O. Networks of the Brain (The MIT Press, 2010).

47. Tijms, B. M., Series, P., Willshaw, D. J. & Lawrie, S. M. Similarity‑based extraction of individual networks from gray matter MRI scans. Cereb. Cortex 22, 1530–1541 (2012).

48. Hagmann, P. et al. Mapping human whole‑brain structural networks with diffusion MRI. PLoS ONE 2, e597 (2007).

49. Bradford, K. et al. Association between early adverse life events and irritable bowel syndrome. Clin. Gastroenterol. Hepatol. 10, 385–390.e1–e3 (2012).

50. Dickhaus, B. et al. Irritable bowel syndrome patients show enhanced modulation of visceral perception by auditory stress. Am. J. Gastroenterol. 98, 135–143 (2003).

51. Labus, J. S. et al. Sex differences in emotion‑related cognitive processes in irritable bowel syndrome and healthy control subjects. Pain 154, 2088–2099 (2013).

52. Tillisch, K. et al. Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology 144, 1394–1401.e1–4 (2013).

53. Tillisch, K., Mayer, E. A. & Labus, J. S. Quantitative meta‑analysis identifies brain regions activated during rectal distension in irritable bowel syndrome. Gastroenterology 140, 91–100 (2011).

54. Farmer, A. D. & Aziz, Q. Visceral pain hypersensitivity in functional gastrointestinal disorders. Br. Med. Bull. 91, 123–136 (2009).

55. Mayer, E. A., Berman, S., Chang, L. & Naliboff, B. D. Sex‑based differences in gastrointestinal pain. Eur. J. Pain 8, 451–463 (2004).

56. Tillisch, K. et al. Sex specific alterations in autonomic function among patients with irritable bowel syndrome. Gut 54, 1396–1401 (2005).

57. Gupta, A. et al. Early adverse life events and resting state neural networks in patients with chronic abdominal pain: evidence for sex differences. Psychosom. Med. 76, 404–412 (2014).

58. Hong, J. Y. et al. Sex and disease‑related alterations of anterior insula functional connectivity in chronic abdominal pain. J. Neurosci. 34, 14252–14259 (2014).

59. Hong, J. Y. et al. Patients with chronic visceral pain show sex‑related alterations in intrinsic oscillations of the resting brain. J. Neurosci. 33, 11994–12002 (2013).

60. Kilpatrick, L. A. et al. Sex‑related differences in prepulse inhibition of startle in irritable bowel syndrome (IBS). Biol. Psychol. 84, 272–278 (2010).

61. Van Oudenhove, L., Vandenberghe, J., Vos, R., Holvoet, L. & Tack, J. Factors associated with co‑morbid irritable bowel syndrome and chronic fatigue‑like symptoms in functional dyspepsia. Neurogastroenterol. Motil. 23, 524–e202 (2011).

62. Naliboff, B. D. et al. Longitudinal change in perceptual and brain activation response to visceral stimuli in irritable bowel syndrome patients. Gastroenterology 131, 352–365 (2006).

63. Naliboff, B. N., Lackner, J. & Mayer, E. A. in Principles of Clinical Gastroenterology (eds Yamada, T. et al.) (Wiley‑Blackwell Publishing 2008).

64. Hubbard, C. S. et al. Corticotropin‑releasing factor receptor 1 antagonist alters regional activation and effective connectivity in an emotional‑arousal circuit during expectation of abdominal pain. J. Neurosci. 31, 12491–12500 (2011).

65. Hall, G. B. et al. Heightened central affective response to visceral sensations of pain and discomfort in IBS. Neurogastroenterol. Motil. 22, 276–e80 (2010).

66. Labus, J. S. et al. Acute tryptophan depletion alters the effective connectivity of emotional arousal circuitry during visceral stimuli in healthy women. Gut 60, 1196–1203 (2011).

67. Straube, T., Schmidt, S., Weiss, T., Mentzel, H. J. & Miltner, W. H. Dynamic activation of the anterior cingulate cortex during anticipatory anxiety. Neuroimage 44, 975–981 (2009).

68. Porro, C. A. et al. Does anticipation of pain affect cortical nociceptive systems? J. Neurosci. 22, 3206–3214 (2002).

69. Koyama, T., McHaffie, J. G., Laurienti, P. J. & Coghill, R. C. The subjective experience of pain: where expectations become reality. Proc. Natl Acad. Sci. USA 102, 12950–12955 (2005).

70. Bornhovd, K. et al. Painful stimuli evoke different stimulus‑response functions in the amygdala, prefrontal, insula and somatosensory cortex: a single‑trial fMRI study. Brain 125, 1326–1336 (2002).

71. Verne, G. N. et al. Central representation of visceral and cutaneous hypersensitivity in the irritable bowel syndrome. Pain 103, 99–110 (2003).

72. Seifert, F. et al. Brain activity during sympathetic response in anticipation and experience of pain. Hum. Brain Mapp. 34, 1768–1782 (2013).

73. Yaguez, L. et al. Brain response to visceral aversive conditioning: a functional magnetic resonance imaging study. Gastroenterology 128, 1819–1829 (2005).

74. Berman, S. M. et al. Reduced brainstem inhibition during anticipated pelvic visceral pain correlates with enhanced brain response to the visceral stimulus in women with irritable bowel syndrome. J. Neurosci. 28, 349–359 (2008).

75. Stephan, E. et al. Functional neuroimaging of gastric distention. J. Gastrointest Surg. 7, 740–749 (2003).

76. Lawal, A. et al. Neurocognitive processing of esophageal central sensitization in the insula and cingulate gyrus. Am. J. Physiol. Gastrointest. Liver Physiol. 294, G787–G794 (2008).

77. Kilpatrick, L. A. et al. The HTR3A polymorphism c. –42C>T is associated with amygdala responsiveness in patients with irritable bowel syndrome. Gastroenterology 140, 1943–1951 (2011).

78. Labus, J. S. et al. Irritable bowel syndrome in female patients is associated with alterations in structural brain networks. Pain 155, 137–149 (2014).

79. de Kloet, E. R., Joels, M. & Holsboer, F. Stress and the brain: from adaptation to disease. Nat. Rev. Neurosci. 6, 463–475 (2005).

80. McEwen, B. S. & Morrison, J. H. The brain on stress: vulnerability and plasticity of the prefrontal cortex over the life course. Neuron 79, 16–29 (2013).

81. Holschneider, D. P., Bradesi, S. & Mayer, E. A. The role of experimental models in developing new treatments for irritable bowel syndrome. Expert Rev. Gastroenterol. Hepatol. 5, 43–57 (2011).

82. Tache, Y., Martinez, V., Wang, L. & Million, M. CRF1 receptor signaling pathways are involved in stress‑related alterations of colonic function and viscerosensitivity: implications for irritable bowel syndrome. Br. J. Pharmacol. 141, 1321–1330 (2004).

83. Labus, J. S. et al. Impaired emotional learning and involvement of the corticotropin‑releasing factor signaling system in patients with irritable bowel syndrome. Gastroenterology 145, 1253–1261 e1–3 (2013).

84. Rubio, A. et al. Uncertainty in anticipation of uncomfortable rectal distension is modulated by the autonomic nervous system—a fMRI study in healthy volunteers. Neuroimage 107C, 110–22 (2014).

85. Kilpatrick, L. A. et al. Alterations in resting state oscillations and connectivity in sensory and motor networks in women with interstitial cystitis/painful bladder syndrome. J. Urol. 192, 947–955 (2014).

86. Apkarian, A. V., Thomas, P. S., Krauss, B. R. & Szeverenyi, N. M. Prefrontal cortical hyperactivity in patients with sympathetically mediated chronic pain. Neurosci. Lett. 311, 193–197 (2001).

REVIEWS

© 2015 Macmillan Publishers Limited. All rights reserved

Page 13: Towards a systems view of IBS - University of Pittsburghpcpr.pitt.edu/wp-content/uploads/2018/01/Mayer-2015.pdf · IBS symptom generation in humans has not firmly been established

604 | OCTOBER 2015 | VOLUME 12 www.nature.com/nrgastro

87. Becerra, L. et al. Intrinsic brain networks normalize with treatment in pediatric complex regional pain syndrome. Neuroimage Clin. 6, 347–369 (2014).

88. DaSilva, A. F. et al. Colocalized structural and functional changes in the cortex of patients with trigeminal neuropathic pain. PLoS ONE 3, e3396 (2008).

89. Ellingson, B. M. et al. Diffusion tensor imaging detects microstructural reorganization in the brain associated with chronic irritable bowel syndrome. Pain 154, 1528–1541 (2013).

90. Jiang, Z. et al. Sex‑related differences of cortical thickness in patients with chronic abdominal pain. PLoS ONE 8, e73932 (2013).

91. Piche, M. et al. Thicker posterior insula is associated with disease duration in women with irritable bowel syndrome (IBS) whereas thicker orbitofrontal cortex predicts reduced pain inhibition in both IBS patients and controls. J. Pain 14, 1217–1226 (2013).

92. Fan, J., McCandliss, B. D., Fossella, J., Flombaum, J. I. & Posner, M. I. The activation of attentional networks. Neuroimage 26, 471–479 (2005).

93. Afzal, M., Potokar, J. P., Probert, C. S. & Munafo, M. R. Selective processing of gastrointestinal symptom‑related stimuli in irritable bowel syndrome. Psychosom. Med. 68, 758–761 (2006).

94. Gibbs‑Gallagher, N. et al. Selective recall of gastrointestinal‑sensation words: evidence for a cognitive‑behavioral contribution to irritable bowel syndrome. Am. J. Gastroenterol. 96, 1133–1138 (2001).

95. Phillips, K., Wright, B. J. & Kent, S. Irritable bowel syndrome and symptom severity: Evidence of negative attention bias, diminished vigour, and autonomic dysregulation. J. Psychosom Res. 77, 13–19 (2014).

96. Tkalcic, M., Domijan, D., Pletikosic, S., Setic, M. & Hauser, G. Attentional biases in irritable bowel syndrome patients. Clin. Res. Hepatol. Gastroenterol. 38, 621–628 (2014).

97. Labus, J. S. et al. Brain networks underlying perceptual habituation to repeated aversive visceral stimuli in patients with irritable bowel syndrome. Neuroimage 47, 952–960 (2009).

98. Blankstein, U., Chen, J., Diamant, N. E. & Davis, K. D. Altered brain structure in irritable bowel syndrome: potential contributions of pre‑existing and disease‑driven factors. Gastroenterology 138, 1783–1789 (2010).

99. Seminowicz, D. A. et al. Cognitive‑behavioral therapy increases prefrontal cortex gray matter in patients with chronic pain. J. Pain 14, 1573–1584 (2013).

100. Elsenbruch, S. et al. Patients with irritable bowel syndrome have altered emotional modulation of neural responses to visceral stimuli. Gastroenterology 139, 1310–1319 (2010).

101. Elsenbruch, S. et al. Affective disturbances modulate the neural processing of visceral pain stimuli in irritable bowel syndrome: an fMRI study. Gut 59, 489–495 (2010).

102. Jui‑Yang Hong et al. IBS patients show altered brain responses during uncertain, but not certain expectation of painful stimulation of the abdominal wall [abstract]. Gastroenterology 148 (Suppl.) S384 (2014).

103. Irwin, M. R. & Cole, S. W. Reciprocal regulation of the neural and innate immune systems. Nat. Rev. Immunol. 11, 625–632 (2011).

104. Lackner, J. M. & Gurtman, M. B. Pain catastrophizing and interpersonal problems: a circumplex analysis of the communal coping model. Pain 110, 597–604 (2004).

105. van Tilburg, M. A., Palsson, O. S. & Whitehead, W. E. Which psychological

factors exacerbate irritable bowel syndrome? Development of a comprehensive model. J. Psychosom Res. 74, 486–492 (2013).

106. Camilleri, M. Genetics of human gastrointestinal sensation. Neurogastroenterol. Motil. 25, 458–466 (2013).

107. Saito, Y. A., Mitra, N. & Mayer, E. A. Genetic approaches to functional gastrointestinal disorders. Gastroenterology 138, 1276–1285 (2010).

108. Gupta, A. et al. Early life adversity: interactions with stress related gene polymorphisms to impact regional brain structure in females. Brain Struct. Funct. http://dx.doi.org/10.1007/s00429‑015‑0996‑9.

109. Kilpatrick, L., Gupta, A., Heendeniya, N., Labus, J. & Mayer, E. A. Corticotropin releasing hormone receptor 1 (crh‑r1) and progestoerone receptor (pgr) polymorphisms interact with early life trauma in healthy controls (hc) and patients with irritable bowel syndrome (IBS) [abstract 666]. Gastroenterology 144 (Suppl. 1), S121 (2013).

110. Gupta, A. et al. Catecholaminergic gene polymorphisms are associated with GI symptoms and morphological brain changes in irritable bowel syndrome. PLoS ONE (in press).

111. Hong, S., Zheng, G. & Wiley, J. W. Epigenetic regulation of genes that modulate chronic stress‑induced visceral pain in the peripheral nervous system. Gastroenterology 148, 148–157.e7 (2015).

112. Tran, L., Schulkin, J., Ligon, C. O. & Greenwood‑Van Meerveld, B. Epigenetic modulation of chronic anxiety and pain by histone deacetylation. Mol. Psychiatry http://dx.doi.org/ 10.1038/mp.2014.122.

113. Videlock, E. J. et al. Childhood trauma is associated with hypothalamic‑pituitary‑adrenal axis responsiveness in irritable bowel syndrome. Gastroenterology 137, 1954–1962 (2009).

114. Heim, C. & Nemeroff, C. B. The role of childhood trauma in the neurobiology of mood and anxiety disorders: preclinical and clinical studies. Biol. Psychiatry 49, 1023–1039 (2001).

115. Klengel, T. & Binder, E. B. Gene‑environment interactions in major depressive disorder. Can. J. Psychiatry 58, 76–83 (2013).

116. Felitti, V. J. et al. Relationship of childhood abuse and household dysfunction to many of the leading causes of death in adults. The Adverse Childhood Experiences (ACE) Study. Am. J. Prev. Med. 14, 245–258 (1998).

117. Berman, S. et al. Evidence for alterations in central noradrenergic signaling in irritable bowel syndrome. Neuroimage 63, 1854–1863 (2012).

118. Chaloner, A., Rao, A., Al‑Chaer, E. D. & Greenwood‑Van Meerveld, B. Importance of neural mechanisms in colonic mucosal and muscular dysfunction in adult rats following neonatal colonic irritation. Int. J. Dev. Neurosci. 28, 99–103 (2010).

119. Flugge, G., van Kampen, M., Meyer, H. & Fuchs, E. α2A and α2C‑adrenoceptor regulation in the brain: α2A changes persist after chronic stress. Eur. J. Neurosci. 17, 917–928 (2003).

120. Chaloner, A. & Greenwood‑Van Meerveld, B. Early life adversity as a risk factor for visceral pain in later life: importance of sex differences. Front. Neurosci. 7, 13 (2013).

121. McEwen, B. S. Understanding the potency of stressful early life experiences on brain and body function. Metabolism 57 (Suppl. 2), S11–S15 (2008).

122. Provencal, N. et al. The signature of maternal rearing in the methylome in rhesus macaque prefrontal cortex and T cells. J. Neurosci. 32, 15626–15642 (2012).

123. Provencal, N. & Binder, E. B. The effects of early life stress on the epigenome: from the womb to

adulthood and even before. Exp. Neurol. 268, 10–20 (2015).

124. Barsky, A. J. & Borus, J. F. Functional somatic syndromes. Ann. Intern. Med. 130, 910–921 (1999).

125. Ahmad, O. F. & Akbar, A. Dietary treatment of irritable bowel syndrome. Br. Med. Bull. 113, 83–90 (2015).

126. Fasano, A., Sapone, A., Zevallos, V. & Schuppan, D. Non‑celiac gluten sensitivity. Gastroenterology 148, 1195–1204 (2015).

127. Lomer, M. C. Review article: the aetiology, diagnosis, mechanisms and clinical evidence for food intolerance. Aliment Pharmacol. Ther. 41, 262–275 (2015).

128. Shepherd, S. J., Halmos, E. & Glance, S. The role of FODMAPs in irritable bowel syndrome. Curr. Opin. Clin. Nutr. Metab. Care 17, 605–609 (2014).

129. Grover, M., Camilleri, M., Smith, K., Linden, D. R. & Farrugia, G. On the fiftieth anniversary. Postinfectious irritable bowel syndrome: mechanisms related to pathogens. Neurogastroenterol. Motil. 26, 156–167 (2014).

130. Spiller, R. & Garsed, K. Postinfectious irritable bowel syndrome. Gastroenterology 136, 1979–1988 (2009).

131. Maxwell, P. R., Rink, E., Kumar, D. & Mendall, M. A. Antibiotics increase functional abdominal symptoms. Am. J. Gastroenterol. 97, 104–108 (2002).

132. Villarreal, A. A., Aberger, F. J., Benrud, R. & Gundrum, J. D. Use of broad‑spectrum antibiotics and the development of irritable bowel syndrome. WMJ 111, 17–20 (2012).

133. Bashashati, M. et al. Cytokine imbalance in irritable bowel syndrome: a systematic review and meta‑analysis. Neurogastroenterol. Motil. 26, 1036–1048 (2014).

134. Darkoh, C. et al. Chemotactic chemokines are important in the pathogenesis of irritable bowel syndrome. PLoS ONE 9, e93144 (2014).

135. Ohman, L. et al. Increased TLR2 expression on blood monocytes in irritable bowel syndrome patients. Eur. J. Gastroenterol. Hepatol. 24, 398–405 (2012).

136. Matricon, J. et al. Review article: associations between immune activation, intestinal permeability and the irritable bowel syndrome. Aliment. Pharmacol. Ther. 36, 1009–1031 (2012).

137. Cremon, C. et al. Mucosal immune activation in irritable bowel syndrome: gender‑dependence and association with digestive symptoms. Am. J. Gastroenterol. 104, 392–400 (2009).

138. Lee, K. J. et al. The alteration of enterochromaffin cell, mast cell, and lamina propria T lymphocyte numbers in irritable bowel syndrome and its relationship with psychological factors. J. Gastroenterol. Hepatol. 23, 1689–1694 (2008).

139. Farhadi, A., Fields, J. Z. & Keshavarzian, A. Mucosal mast cells are pivotal elements in inflammatory bowel disease that connect the dots: stress, intestinal hyperpermeability and inflammation. World J. Gastroenterol. 13, 3027–3030 (2007).

140. Cole, S. W. Social regulation of human gene expression. Curr. Dir. Psychol. Sci. 18, 132–137 (2009).

141. Miller, G. E. et al. Low early‑life social class leaves a biological residue manifested by decreased glucocorticoid and increased proinflammatory signaling. Proc. Natl Acad. Sci. USA 106, 14716–14721 (2009).

142. Szyf, M. The early life environment and the epigenome. Biochim. Biophys. Acta 1790, 878–885 (2009).

REVIEWS

© 2015 Macmillan Publishers Limited. All rights reserved

Page 14: Towards a systems view of IBS - University of Pittsburghpcpr.pitt.edu/wp-content/uploads/2018/01/Mayer-2015.pdf · IBS symptom generation in humans has not firmly been established

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 12 | OCTOBER 2015 | 605

143. Gupta, A. et al. Early adverse life events: influence on resting state connectivity in patients with chronic abdominal pain. Psychosom. Med. 76, 404–412 (2014).

144. Cole, S. W. Human social genomics. PLoS Genet. 10, e1004601 (2014).

145. Cole, S. W. et al. Transcriptional modulation of the developing immune system by early life social adversity. Proc. Natl Acad. Sci. USA 109, 20578–20583 (2012).

146. Powell, N. D. et al. Social stress up‑regulates inflammatory gene expression in the leukocyte transcriptome via beta‑adrenergic induction of myelopoiesis. Proc. Natl Acad. Sci. USA 110, 16574–16579 (2013).

147. Wohleb, E. S., Powell, N. D., Godbout, J. P. & Sheridan, J. F. Stress‑induced recruitment of bone marrow‑derived monocytes to the brain promotes anxiety‑like behavior. J. Neurosci. 33, 13820–13833 (2013).

148. Wohleb, E. S. et al. Re‑establishment of anxiety in stress‑sensitized mice is caused by monocyte trafficking from the spleen to the brain. Biol. Psychiatry 75, 970–981 (2014).

149. Wohleb, E. S. et al. beta‑Adrenergic receptor antagonism prevents anxiety‑like behavior and microglial reactivity induced by repeated social defeat. J. Neurosci. 31, 6277–6288 (2011).

150. Tillisch, K. et al. Sex specific alterations in autonomic function among patients with irritable bowel syndrome. Gut 54, 1396–1401 (2005).

151. Berman, S. et al. Differential noradrenergic (NE) modulation of brain metabolic and electrical activity in IBS patients and healthy controls. Gastroenterology 132, A726 (2007).

152. Labus, J. et al. Sex‑specific differences in a brain network functioning during anticipation of rectal discomfort in irritable bowel syndrome patients (IBS). Gastroenterology 130, A93 (2006).

153. Mayer, E. A. et al. Correlation between gene expression profiles in peripheral blood mononuclear cells and structural and functional brain networks in chronic visceral pain. Neuropsychopharmacology 39, S641 (2014).

154. Cole, S. W., Hawkley, L. C., Arevalo, J. M. & Cacioppo, J. T. Transcript origin analysis identifies antigen‑presenting cells as primary targets of socially regulated gene expression in leukocytes. Proc. Natl Acad. Sci. USA 108, 3080–3085 (2011).

155. Beissner, F., Meissner, K., Bar, K. J. & Napadow, V. The autonomic brain: an activation likelihood estimation meta‑analysis for central processing of autonomic function. J. Neurosci. 33, 10503–10511 (2013).

156. Seminowicz, D. A. et al. Regional gray matter density changes in brains of patients with irritable bowel syndrome. Gastroenterology 139, U48–U82 (2010).

157. Cryan, J. F. & Dinan, T. G. Mind‑altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci. 13, 701–712 (2012).

158. Mayer, E. A. & Tillisch, K. Gut brain axis and the microbiome. J. Clin. Invest. 125, 926–938 (2015). 

159. Stilling, R. M., Dinan, T. G. & Cryan, J. F. Microbial genes, brain & behaviour—epigenetic regulation of the gut‑brain axis. Genes Brain Behav. 13, 69–86 (2014).

160. Rhee, S. H., Pothoulakis, C. & Mayer, E. A. Principles and clinical implications of the brain‑gut‑enteric microbiota axis. Nat. Rev. Gastroenterol. Hepatol. 6, 306–314 (2009).

161. Benton, D., Williams, C. & Brown, A. Impact of consuming a milk drink containing a probiotic on mood and cognition. Eur. J. Clin. Nutr. 61, 355–361 (2007).

162. Messaoudi, M. et al. Beneficial psychological effects of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in healthy human volunteers. Gut Microbes 2, 256–261 (2011).

163. McNulty, N. P. et al. The impact of a consortium of fermented milk strains on the gut microbiome of gnotobiotic mice and monozygotic twins. Sci. Transl. Med. 3, 106ra106 (2011).

164. Mayer, E. A. et al. Differences in brain responses to visceral pain between patients with irritable bowel syndrome and ulcerative colitis. Pain 115, 398–409 (2005).

165. Hood, L., Heath, J. R., Phelps, M. E. & Lin, B. Systems biology and new technologies enable predictive and preventative medicine. Science 306, 640–643 (2004).

166. Hornberg, J. J., Bruggeman, F. J., Westerhoff, H. V. & Lankelma, J. Cancer: a systems biology disease. Biosystems 83, 81–90 (2006).

167. Likic, V. A., McConville, M. J., Lithgow, T. & Bacic, A. Systems biology: the next frontier for bioinformatics. Adv. Bioinformatics 2010, 268925 (2010).

168. Chang, L., Lembo, A. & Sultan, S. American Gastroenterological Association institute technical review on the pharmacological management of irritable bowel syndrome. Gastroenterology 147, 1149–1172.e2 (2014).

169. Mayer, E. A. & Collins, S. M. Evolving pathophysiologic models of functional gastrointestinal disorders. Gastroenterology 122, 2032–2048 (2002).

170. Tache, Y., Martinez, V., Million, M. & Wang, L. Stress and the gastrointestinal tract III. Stress‑related alterations of gut motor function: role of brain corticotropin‑releasing factor receptors. Am. J. Physiol. Gastrointest. Liver Physiol. 280, G173–177 (2001).

171. Patel, V. R., Eckel‑Mahan, K., Sassone‑Corsi, P. & Baldi, P. CircadiOmics: integrating circadian genomics, transcriptomics, proteomics and metabolomics. Nat, Methods 9, 772–773 (2012).

172. Patel, V. R., Eckel‑Mahan, K., Sassone‑Corsi, P. & Baldi, P. How pervasive are circadian oscillations? Trends Cell Biol. 24, 329–331 (2014).

173. Partch, C. L., Green, C. B. & Takahashi, J. S. Molecular architecture of the mammalian circadian clock. Trends Cell Biol. 24, 90–99 (2014).

174. Eckel‑Mahan, K. L. et al. Reprogramming of the circadian clock by nutritional challenge. Cell 155, 1464–1478 (2013).

175. Takahashi, J. S., Hong, H. K., Ko, C. H. & McDearmon, E. L. The genetics of mammalian circadian order and disorder: implications for physiology and disease. Nat. Rev. Genet. 9, 764–775 (2008).

176. Bellet, M. M. et al. Circadian clock regulates the host response to Salmonella. Proc. Natl Acad. Sci. USA 110, 9897–9902 (2013).

177. Zhang, R., Lahens, N. F., Ballance, H. I., Hughes, M. E. & Hogenesch, J. B. A circadian gene expression atlas in mammals: implications for biology and medicine. Proc. Natl Acad. Sci. USA 111, 16219–16224 (2014).

178. Bonaz, B. L. & Bernstein, C. N. Brain‑gut interactions in inflammatory bowel disease. Gastroenterology 144, 36–49 (2013).

179. Greicius, M. D., Krasnow, B., Reiss, A. L. & Menon, V. Functional connectivity in the resting brain: a network analysis of the default mode hypothesis. Proc. Natl Acad. Sci. USA 100, 253–258 (2003).

180. Buckner, R. L., Andrews‑Hanna, J. R. & Schacter, D. L. The brain’s default network: anatomy, function, and relevance to disease. Ann. N. Y. Acad. Sci. 1124, 1–38 (2008).

181. Raichle, M. E. et al. A default mode of brain function. Proc. Natl Acad. Sci. USA 98, 676–682 (2001).

182. Qin, P. & Northoff, G. How is our self related to midline regions and the default‑mode network? Neuroimage 57, 1221–1233 (2011).

183. Damoiseaux, J. S. et al. Consistent resting‑state networks across healthy subjects. Proc. Natl Acad. Sci. USA 103, 13848–13853 (2006).

184. Labus, J. S. et al. Sex differences in brain activity during aversive visceral stimulation and its expectation in patients with chronic abdominal pain: a network analysis. Neuroimage 41, 1032–1043 (2008).

185. Pezawas, L. et al. 5‑HTTLPR polymorphism impacts human cingulate‑amygdala interactions: a genetic susceptibility mechanism for depression. Nat. Neurosci. 8, 828–834 (2005).

186. Stein, J. L. et al. A validated network of effective amygdala connectivity. Neuroimage 36, 736–745 (2007).

187. Critchley, H. D., Nagai, Y., Gray, M. A. & Mathias, C. J. Dissecting axes of autonomic control in humans: Insights from neuroimaging. Auton. Neurosci. 161, 34–42 (2011).

188. Saper, C. B. The central autonomic nervous system: conscious visceral perception and autonomic pattern generation. Annu. Rev. Neurosci. 25, 433–469 (2002).

189. Biswal, B., Yetkin, F. Z., Haughton, V. M. & Hyde, J. S. Functional connectivity in the motor cortex of resting human brain using echo‑planar MRI. Magn. Reson. Med. 34, 537–541 (1995).

190. Jones, L. M., Fontanini, A., Sadacca, B. F., Miller, P. & Katz, D. B. Natural stimuli evoke dynamic sequences of states in sensory cortical ensembles. Proc. Natl Acad. Sci. USA 104, 18772–18777 (2007).

191. Alcauter, S. et al. Development of thalamocortical connectivity during infancy and its cognitive correlations. J. Neurosci. 34, 9067–9075 (2014).

192. Levinthal, D. J. & Strick, P. L. The motor cortex communicates with the kidney. J. Neurosci. 32, 6726–6731 (2012).

193. Menon, V. Large‑scale brain networks and psychopathology: a unifying triple network model. Trends Cogn. Sci. 15, 483–506 (2011).

194. Borsook, D., Edwards, R., Elman, I., Becerra, L. & Levine, J. Pain and analgesia: the value of salience circuits. Prog. Neurobiol. 104, 93–105 (2013).

195. Greicius, M. D. et al. Resting‑state functional connectivity in major depression: abnormally increased contributions from subgenual cingulate cortex and thalamus. Biol. Psychiatry 62, 429–437 (2007).

196. Laird, A. R. et al. Behavioral interpretations of intrinsic connectivity networks. J. Cogn. Neurosci. 23, 4022–4037 (2011).

197. Grupe, D. W. & Nitschke, J. B. Uncertainty and anticipation in anxiety: an integrated neurobiological and psychological perspective. Nat. Rev. Neurosci. 14, 488–501 (2013).

AcknowledgementsSupported by funding National Institutes of Health grants P50 DK064539 (E.A.M.), R01 DK048351 (E.A.M.) and P30 DK041301 (E.R.). The authors thank Cathy Liu and Arpana Gupta for invaluable editorial assistance.

Author contributionsThe article was conceptualized and composed by E.A.M. Inputs to the following sections came from the following co‑authors: immune system (S.W.C.); brain networks (J.S.L.); gut microbiota (K.T.); and systems biology (P.B.).

REVIEWS

© 2015 Macmillan Publishers Limited. All rights reserved